MedPath

Glenzocimab Advanced Drug Monograph

Published:Jun 13, 2025

Generic Name

Glenzocimab

Drug Type

Biotech

CAS Number

2101829-58-5

Glenzocimab (DB16175): A Comprehensive Review of a Glycoprotein VI Inhibitor for Acute Ischemic Stroke

I. Introduction to Glenzocimab

A. Overview and Rationale for Development

Glenzocimab, also known by its developmental identifier ACT017, is an investigational biopharmaceutical agent that has been primarily developed as an antiplatelet therapy. Its main therapeutic target has been cardiovascular emergencies, with a specific and significant focus on the acute phase of ischemic stroke (AIS).[1] The fundamental rationale for the development of glenzocimab is rooted in the critical unmet need for effective and safe antithrombotic treatments that can be administered rapidly during the hyperacute phase of AIS. The primary goals of such an intervention are to halt the progression of the occlusive thrombus, prevent further thromboembolic events, and improve microcirculatory reperfusion, ideally with a superior safety profile, particularly a diminished risk of bleeding complications when compared with existing antithrombotic or thrombolytic therapies.[1] Glenzocimab was conceived to address the crucial therapeutic window in the initial hours immediately following a stroke, a period where timely and effective intervention is paramount to minimizing irreversible neurological damage.[1]

B. Chemical and Physical Properties

Glenzocimab is identified by the DrugBank ID DB16175 and CAS Number 2101829-58-5.[5] It is classified as a biotechnology-derived product. More specifically, glenzocimab is a humanized monoclonal antibody fragment (Fab).[1] This structural characteristic is significant, as Fab fragments typically exhibit different pharmacokinetic profiles, such as shorter half-lives and potentially reduced immunogenicity, compared to full-length monoclonal antibodies. The molecular weight of glenzocimab is approximately 48 kDa [1], a size consistent with a Fab fragment and notably below the renal glomerular filtration threshold, which suggests a potential route of renal clearance. While one source mentions a molecular weight of 145.5 KD [5], this likely refers to the parent full antibody, as multiple sources confirm glenzocimab's nature as a Fab fragment.[1] Glenzocimab was derived from the murine monoclonal antibody 9O12 through humanization processes, a standard technique to minimize potential immunogenic responses in human subjects.[4] For clinical administration, glenzocimab is formulated for intravenous infusion.[1]

The development of a Fab fragment like glenzocimab, rather than a full monoclonal antibody, represents a strategic choice for an agent intended for acute cardiovascular emergencies. Fab fragments, due to their smaller size, may offer advantages such as enhanced tissue penetration, potentially allowing for more rapid access to the site of a thrombus. Furthermore, they typically possess a shorter plasma half-life (glenzocimab's terminal half-life is reported as 9.6 hours [1]), which can be beneficial in acute settings. A shorter duration of action allows for a quicker cessation of the drug's effect if adverse events, such as bleeding, occur, or if emergency surgical intervention becomes necessary. This contrasts with intact immunoglobulins (IgGs), which generally have much longer half-lives due to FcRn-mediated recycling mechanisms. The primary focus on AIS, a condition characterized by a narrow therapeutic window and a significant unmet need for safer adjunctive therapies, highlights the perceived novelty and potential safety advantages associated with glenzocimab's specific molecular target and mechanism of action.[2]

Table 1: Key Identifiers and Properties of Glenzocimab

PropertyDetailReference(s)
DrugBank IDDB16175
CAS Number2101829-58-55
TypeBiotech; Humanized monoclonal antibody fragment (Fab)1
Molecular TargetPlatelet Glycoprotein VI (GPVI)1
Molecular WeightApprox. 48 kDa1
DeveloperActicor Biotech2
OriginDerived from murine monoclonal antibody 9O124
FormulationIntravenous infusion1

II. Mechanism of Action

A. Target: Platelet Glycoprotein VI (GPVI)

Glenzocimab exerts its pharmacological effect by specifically binding to human platelet glycoprotein VI (GPVI).[1] GPVI is a type I transmembrane glycoprotein that is uniquely expressed on the surface of megakaryocytes and mature circulating platelets.[4] It functions as a critical signaling receptor involved in the initial stages of hemostasis and thrombosis. GPVI's primary physiological ligands are collagen, exposed at sites of vascular injury, and fibrin, a key component of formed thrombi. The interaction of GPVI with these ligands initiates platelet adhesion, activation, aggregation, and degranulation, processes central to thrombus formation and stabilization.[1] The expression level of GPVI on platelets is moderate, estimated at 4000 to 6000 copies per platelet, a density considered amenable to pharmacological saturation by an antagonistic agent.[4]

B. Molecular Interactions and Inhibition of Platelet Aggregation

Glenzocimab binds with high affinity to the D2 domain of GPVI, with a reported dissociation constant (Kd​) in the nanomolar range (e.g., 4.1 nM).[1] Crystallographic studies have provided detailed insights into this interaction, revealing that glenzocimab binding to the D2 domain of GPVI sterically hinders homotypic interactions between GPVI molecules. This interference prevents the formation of GPVI dimers on the platelet surface.[4] The dimerization of GPVI is understood to be a crucial step for its high-affinity binding to ligands such as collagen and fibrin, and subsequent signal transduction leading to platelet activation.

The inhibitory mechanism of glenzocimab is not solely based on steric hindrance; it also involves the induction of conformational changes within the GPVI molecule upon Fab binding.[4] This combination of steric blockade of dimerization and allosteric modulation of GPVI structure effectively prevents the glycoprotein from productively interacting with its major physiological ligands, collagen and fibrin. As a direct consequence, glenzocimab potently inhibits collagen-induced platelet aggregation and degranulation

in vitro, with a reported half-maximal inhibitory concentration (IC50) of 3.2 μg/mL for collagen-induced platelet aggregation.[1]

C. Impact on Thrombus Formation, Stability, and Thrombo-inflammation

By effectively blocking GPVI activation, glenzocimab is designed to impede the growth of existing thrombi and inhibit the formation of new clots. This is particularly relevant in the context of microvascular occlusions that can occur downstream of a primary arterial blockage in AIS.[1] There is also evidence suggesting that glenzocimab may promote the disaggregation of platelets from an established thrombus.[2]

A significant and differentiating aspect of glenzocimab's mechanism of action is its potential to inhibit thrombo-inflammatory processes.[1] Thrombo-inflammation involves complex interplay between activated platelets and immune cells, such as neutrophils. These interactions contribute to the inflammatory cascade and secondary tissue damage observed in ischemic events like stroke. By modulating GPVI-mediated platelet activity, glenzocimab may attenuate these deleterious inflammatory responses.

The unique mode of action of glenzocimab, primarily targeting GPVI dimerization and inducing conformational changes rather than directly competing with ligands at their principal binding sites on GPVI [4], may underpin its observed favorable bleeding profile. Pathological thrombus formation often involves specific patterns of GPVI clustering and activation that could be more susceptible to the disruption of dimerization than the processes involved in physiological hemostasis. Physiological hemostasis typically occurs under conditions of high shear stress and massive collagen exposure at a site of significant vascular injury. If glenzocimab's action subtly modulates GPVI to prevent pathological hyperactivation while permitting some level of basal function, it might preserve essential hemostatic plug formation. This hypothesis is supported by preclinical findings that demonstrate antithrombotic effects without a significant increase in bleeding time.[1]

Furthermore, the capacity of glenzocimab to interfere with GPVI-fibrin interactions [4] is particularly pertinent to thrombus stability and growth. While collagen typically initiates platelet activation at sites of vascular injury, fibrin forms the structural meshwork that consolidates the growing thrombus. An agent that disrupts platelet interaction with this fibrin scaffold could limit thrombus maturation and potentially render it more susceptible to endogenous or exogenous lytic therapies.

The targeting of thrombo-inflammation [1] offers a therapeutic dimension beyond simple antithrombosis. In the context of ischemic stroke, post-ischemic inflammation significantly contributes to secondary neuronal injury. Platelets are active participants in orchestrating these inflammatory responses. By inhibiting GPVI, which is implicated in platelet-leukocyte interactions, glenzocimab might mitigate the extent of this secondary damage, an effect not typically addressed by conventional antiplatelet agents focusing solely on aggregation.

III. Pharmacokinetics and Pharmacodynamics (PK/PD)

A. Absorption, Distribution, Metabolism, and Excretion (ADME)

Glenzocimab is administered via intravenous infusion, as established in its clinical development program.[1] Population pharmacokinetic (PK) modeling from a Phase 1 study in healthy volunteers indicated that glenzocimab follows a two-compartment model with dose-proportional kinetics.[1] The central volume of distribution (

Vc​) was estimated to be 4.1 liters.[1] As a humanized Fab fragment, glenzocimab is expected to be metabolized through general protein catabolism into smaller peptides and amino acids, which are then recycled or excreted.

The elimination of glenzocimab is biphasic. It exhibits a rapid initial distribution phase with a half-life (t1/2α​) of 0.84 hours, followed by a slower terminal elimination phase with a half-life (t1/2β​) of 9.6 hours.[1] Considering its molecular weight of approximately 48 kDa [1], which is below the glomerular filtration threshold of about 70 kDa, renal clearance was anticipated to be a significant elimination pathway. However, PK/PD simulations revealed that variations in creatinine clearance had a limited impact on overall glenzocimab exposure. This finding suggests that dose adjustments based on renal function might not be necessary, a significant advantage in acute care settings.[1] The potential role of target-mediated drug disposition (TMDD), where the drug is eliminated via binding to its target receptor (GPVI on platelets) followed by internalization and degradation, was considered during modeling but was not definitively established as the predominant clearance mechanism in the available information.[1]

B. Dose-Response Relationship and Inhibition of Platelet Aggregation

Glenzocimab demonstrates a clear dose-dependent inhibitory effect on ex vivo collagen-induced platelet aggregation.[1] The pharmacodynamic (PD) effect was characterized using an immediate effect model in population PK/PD analyses. A particularly interesting finding from the Phase 1 study was a dose-dependent half-maximal inhibitory concentration (IC50); specifically, higher administered doses of glenzocimab resulted in a more potent antiplatelet effect even at the same measured plasma concentrations of the drug. The precise mechanism underlying this "overproportional concentration effect" at higher doses was not fully elucidated by the initial modeling efforts.[1]

In vitro, the IC50 for glenzocimab's inhibition of collagen-induced platelet aggregation has been reported as 3.2 μg/mL.[1]

C. Population PK/PD Modeling Insights and Predictions

Based on the integrated PK/PD model developed from Phase 1 data, simulations were performed to predict the antiplatelet effect under clinical dosing scenarios. These simulations predicted that a 1000 mg dose of glenzocimab, administered as a 6-hour intravenous infusion, would achieve a reduction in platelet aggregation to 20% of baseline levels in 100% of subjects by the end of the 6-hour infusion. Furthermore, this level of inhibition was predicted to be maintained in 60% of subjects at 12 hours after the initiation of dosing.[1] A key therapeutic consideration guiding the desired PK/PD profile was that the significant antiplatelet effect of glenzocimab should ideally not extend beyond 24 hours. This duration was chosen to minimize potential overlap and synergistic bleeding risks if other anticoagulant or antiplatelet therapies were to be initiated subsequently.[1]

The observed terminal half-life of approximately 9.6 hours [1] is relatively short for a biologic therapeutic, a characteristic that is generally advantageous for an agent intended for acute stroke. This allows for a reasonably rapid decline in drug levels and offset of action if bleeding complications were to occur or if urgent surgical intervention became necessary post-administration. This pharmacokinetic profile aligns well with the therapeutic goal of an effect primarily limited to the acute phase of stroke, roughly within 24 hours.[1] In the dynamic and often unpredictable clinical course of acute stroke, a drug with a shorter half-life offers clinicians greater flexibility and a quicker return to baseline hemostatic function if the drug's effects need to be curtailed, thereby reducing the window of risk for bleeding complications compared to antiplatelet agents with much longer durations of action.

The "overproportional concentration effect at higher doses" [1] noted in early studies is a complex pharmacodynamic feature that merits further investigation. This phenomenon, where the drug becomes unexpectedly more potent per unit of concentration as the dose increases, deviates from simple receptor occupancy models. It could suggest several underlying mechanisms, such as the saturation of a plasma binding protein that limits the availability of free drug at lower concentrations, the engagement of a secondary, higher-threshold inhibitory mechanism at elevated concentrations, or complex cooperative binding or receptor clustering dynamics on the platelet surface. A thorough understanding of this effect would be crucial for precise dose optimization, predicting inter-individual variability in response, and fully characterizing the drug's therapeutic window.

The finding that renal function appears to have a limited impact on glenzocimab exposure, as suggested by simulations [1], carries significant clinical advantages, particularly in the emergency management of acute stroke. Patients presenting with acute stroke, especially elderly individuals, frequently have co-existing conditions, including varying degrees of renal impairment. The ability to administer a standard dose of glenzocimab without the immediate need for renal function assessment and dose adjustment simplifies treatment protocols in time-sensitive situations, reduces the potential for dosing errors, and allows for more rapid initiation of therapy, which is of paramount importance in the hyperacute phase of stroke care.

IV. Preclinical Development

A. Efficacy in Animal Models of Thrombosis

The antithrombotic potential of glenzocimab, and its parent murine Fab 9O12, was consistently demonstrated in a range of experimental animal models of thrombosis.[2] These preclinical studies were crucial in establishing the proof-of-concept for GPVI inhibition as a therapeutic strategy. Notably, glenzocimab provided protection against thrombus formation in humanized GPVI mouse models, where mouse platelets express human GPVI, as well as in non-human primate models of thrombosis.[4] These models aim to mimic aspects of human thrombotic disease and provide a translational bridge to clinical studies.

B. Assessment of Bleeding Risk

A defining characteristic and a key anticipated advantage of glenzocimab, highlighted throughout its preclinical development, was its ability to inhibit thrombosis without inducing a corresponding significant increase in bleeding time or the incidence of pathological bleeding.[1] Studies conducted in cynomolgus monkeys showed that administration of glenzocimab resulted in efficient and reversible inhibition of GPVI-mediated platelet function, yet did not adversely affect systemic platelet counts or prolong standardized bleeding times.[1]

More recent and particularly relevant preclinical investigations focused on the safety of glenzocimab in the context of intracranial hemorrhage (ICH). Using mouse models of ICH, including collagenase-induced striatal hemorrhage and hyperglycemia-induced hemorrhagic transformation of an ischemic stroke, researchers found that glenzocimab administration did not exacerbate bleeding severity or hematoma volume. This was in stark contrast to the GPIIb/IIIa inhibitor eptifibatide, a potent antiplatelet agent, which significantly increased intracranial bleeding in the same experimental models.[9]

The consistent preclinical observation that glenzocimab could achieve antithrombotic efficacy without a corresponding increase in bleeding risk [1] formed the most compelling scientific rationale for advancing the drug into clinical trials for AIS. In AIS, the risk of hemorrhagic complications, particularly ICH, is a major limitation for existing antithrombotic and thrombolytic therapies. A novel agent that could effectively decouple antithrombotic efficacy from bleeding propensity would represent a significant paradigm shift in stroke treatment.

The specific demonstration of safety in preclinical ICH models [9] is particularly significant. This finding suggested a unique safety margin that could, in theory, allow for the administration of glenzocimab even before neuroimaging definitively confirms the ischemic nature of the stroke and rules out primary hemorrhage. Current standard AIS treatment protocols mandate urgent neuroimaging (typically CT scan) to exclude ICH before any thrombolytic or potent antiplatelet therapy can be initiated. This "door-to-imaging" and "imaging-to-needle" time contributes to treatment delays. If glenzocimab were indeed proven safe in the presence of ICH, it could potentially be administered by first responders in the pre-hospital setting or immediately upon emergency department arrival, thereby dramatically reducing time-to-treatment and potentially improving neurological outcomes by targeting the ischemic penumbra at an earlier stage. This represents a major potential advantage over existing therapies.

V. Clinical Development in Acute Ischemic Stroke

A. Rationale for Glenzocimab in AIS

The primary rationale for investigating glenzocimab in acute ischemic stroke was derived from its novel mechanism of action, targeting GPVI-mediated platelet aggregation and thrombus formation, combined with its promising preclinical safety profile, which consistently indicated a low propensity to cause bleeding.[1] Glenzocimab was developed as an adjunctive therapy to be administered alongside the existing standard of care for AIS. This standard care typically includes intravenous thrombolysis (IVT) with alteplase, and/or mechanical thrombectomy (MT) for eligible patients with large vessel occlusion.[2] The intended role of glenzocimab was to enhance the quality of reperfusion achieved by these primary treatments and to prevent early reocclusion or further thromboembolic events in the microvasculature.

B. Phase 1b/2a Study (ACTIMIS - NCT03803007)

The ACTIMIS trial was a randomized, double-blind, placebo-controlled study designed to evaluate the safety and tolerability of glenzocimab in AIS patients. The Phase 1b component involved dose-escalation, testing doses of 125 mg, 250 mg, 500 mg, and 1000 mg of glenzocimab, or placebo, with approximately 12 patients per group. From this phase, the 1000 mg dose was selected as the recommended dose for further evaluation in Phase 2a.[2] The Phase 2a part of the study enrolled 106 patients with AIS who were eligible for treatment with alteplase, with or without subsequent mechanical thrombectomy; 53 patients received 1000 mg glenzocimab and 53 received placebo.[6]

The primary endpoint of the ACTIMIS study was safety. Glenzocimab, at the 1000 mg dose, was found to be well tolerated.[2] A key safety finding was a notable reduction in the incidence of intracranial hemorrhage. Symptomatic ICH occurred in 0% of patients treated with 1000 mg glenzocimab compared to 10% (5 patients) in the placebo group.[6] Furthermore, non-symptomatic hemorrhagic transformation was observed in 31% of glenzocimab-treated patients versus 50% in the placebo group.[6]

Although ACTIMIS was primarily a safety study and underpowered for definitive efficacy assessment, several positive exploratory efficacy signals were observed. There was a reduction in all-cause mortality in the glenzocimab 1000 mg group (7%, 4 patients) compared to the placebo group (21%, 11 patients).[6] Post-hoc analyses of brain imaging data, utilizing artificial intelligence, suggested a reduction in both the number and volume of intracerebral lesions in patients who received glenzocimab.[11] The positive results from the ACTIMIS study were published in The Lancet Neurology in January 2024.[11]

C. Phase 2/3 Study (ACTISAVE - NCT05070260)

Following the encouraging results from ACTIMIS, the ACTISAVE trial was initiated. This was an international, adaptive, multicenter, randomized, double-blind, placebo-controlled, parallel-group Phase 2/3 study.[2] A total of 436 AIS patients were randomized to receive a single 1000 mg dose of glenzocimab or placebo. The study drug was administered as an add-on to the standard of care (thrombolysis with or without thrombectomy) within 4.5 hours of stroke symptom onset.[2] The trial was conducted across centers in the USA, European Union (including France, Germany, Belgium, Spain, Slovakia, Denmark, Czech Republic), the United Kingdom, and Israel.[8]

The primary efficacy endpoint for ACTISAVE was the proportion of patients experiencing severe disability or death, defined as a modified Rankin Scale (mRS) score of 4-6, at 90 days post-stroke.[11] The results of the ACTISAVE trial, presented at the European Stroke Organisation Conference (ESOC) in May 2024, indicated that the study

did not meet its primary endpoint. Glenzocimab did not demonstrate a statistically significant reduction in the proportion of patients with an mRS score of 4-6 at 90 days in the overall study population.[11]

However, a trend towards benefit was observed in a pre-specified analysis focusing on the proportion of patients achieving an mRS score of 0-1 (indicating no or minimal disability, often termed "return to normal life"), an endpoint also recommended by the FDA. This positive trend was particularly noted in the subgroup of patients who achieved complete recanalization (eTICI score 3) following mechanical thrombectomy.[11] Post-hoc analyses, which attempted to adjust for baseline patient heterogeneity and imbalances (e.g., in patients who received mechanical thrombectomy in addition to thrombolysis, those on concomitant antithrombotic medications, or those with high NIHSS scores at inclusion), suggested a more favorable outcome for glenzocimab concerning the mRS 0-1 score.[14] Despite the primary endpoint miss, the favorable safety profile of glenzocimab was reaffirmed in the ACTISAVE trial.[11]

D. Phase 2/3 Study (GREEN - NCT05559398)

The GREEN trial was another Phase 2/3 study, designed as a randomized, double-blind, multicenter, placebo-controlled investigation. Its objective was to evaluate the efficacy and safety of glenzocimab when used as an adjunct to mechanical thrombectomy alone in AIS patients presenting within 24 hours of symptom onset. This study was part of the RHU BOOSTER program.[2]

The GREEN study was discontinued prematurely. This decision was based on the recommendation of the Independent Monitoring Committee (IMC) following a planned futility analysis. The analysis, which included data from the first 78 of the 108 patients enrolled at that time, indicated that the trial was unlikely to meet its efficacy objectives. Consequently, the sponsor, Assistance Publique Hôpitaux de Paris (APHP), in agreement with the principal investigator, decided to halt further enrolment.[2]

Table 2: Summary of Key Glenzocimab Clinical Trials in Acute Ischemic Stroke

FeatureACTIMIS (NCT03803007)ACTISAVE (NCT05070260)GREEN (NCT05559398)
Phase1b/2a2/3 (Adaptive)2/3
Patient PopulationAIS, eligible for IVT +/- MTAIS, eligible for IVT +/- MT, within 4.5 hrs of onsetAIS, eligible for MT, within 24 hrs of onset
N (Glenzo/Placebo)Ph1b: 60 total; Ph2a: 53/53218/218 (Total N=436)108 enrolled (futility on first 78)
InterventionGlenzocimab 125-1000mg IV or PlaceboGlenzocimab 1000mg IV or PlaceboGlenzocimab 1000mg IV or Placebo
Primary EndpointSafety & TolerabilityProportion mRS 4-6 at 90 daysEfficacy (functional outcome, mRS) at 90 days
Primary Efficacy Met?N/A (Safety primary)NoN/A (Halted for futility)
mRS 0-1 OutcomeNot primary; positive imaging trendsTrend towards benefit, esp. in eTICI 3 subgroup; more favorable in post-hoc analysesNot available (halted)
mRS 4-6 OutcomeNot primary; mortality lower (7% vs 21%)No significant differenceNot available (halted)
sICH Rate0% (Glenzocimab) vs 10% (Placebo) in Ph2aFavorable safety profile confirmedNot fully analyzed (halted)
Mortality (All-Cause)7% (Glenzocimab) vs 21% (Placebo) in Ph2aNot specified as significantly different in snippetsNot available (halted)
Status/PublicationCompleted; Published Lancet Neurology Jan 2024Completed; Results ESOC May 2024; Did not meet primary endpointDiscontinued due to futility (July 2024)

The trajectory of glenzocimab's clinical development in AIS illustrates a common narrative in pharmaceutical research: initial promise often faces significant hurdles in later-stage, larger trials. The encouraging safety signals from ACTIMIS, particularly the observed reduction in sICH and mortality compared to placebo [6], provided a strong impetus for further investigation. However, the failure of the larger, pivotal ACTISAVE trial to meet its primary efficacy endpoint concerning severe disability or death (mRS 4-6) [11] represents a substantial setback. This discrepancy underscores the inherent difficulties in translating positive signals from smaller, early-phase studies into definitive efficacy in more heterogeneous late-phase populations, a challenge frequently encountered in the complex field of stroke drug development.

Nevertheless, the consistent observation of a trend towards benefit in achieving an excellent functional outcome (mRS 0-1, return to normal life) in the ACTISAVE trial, especially within the subgroup of patients who achieved complete recanalization after thrombectomy (eTICI score 3) [11], suggests that glenzocimab's therapeutic effect might be more nuanced. It is plausible that its primary benefit lies in optimizing the quality of reperfusion or preventing microvascular re-occlusion in patients who have already achieved successful macrovascular recanalization. If this is the case, the drug's impact might be more evident in facilitating shifts from mild or moderate disability to no or minimal disability (thus improving mRS 0-1 rates) rather than preventing severe disability or death across a broad AIS population that includes many patients with poor prognostic factors irrespective of adjunctive therapy. The FDA's recommendation of mRS 0-1 as a relevant endpoint [14] further lends credence to the importance of this observation.

The subsequent discontinuation of the GREEN trial due to futility [13] compounded the challenges. These clinical trial outcomes almost certainly created significant financial pressure on Acticor Biotech, directly contributing to the company's eventual liquidation proceedings.[3] This sequence highlights the critical dependence of clinical-stage biopharmaceutical companies on successful trial outcomes for continued funding and viability.

Despite the efficacy challenges in broad AIS populations, the consistently favorable safety profile of glenzocimab, particularly the low rates of sICH observed in ACTIMIS [6] and the good overall tolerability in ACTISAVE [11], remains a notable and potentially valuable attribute. For any antiplatelet or antithrombotic agent developed for AIS, where bleeding risk is a primary concern, such a safety profile is highly desirable.

VI. Clinical Development in Other Cardiovascular Indications

A. Myocardial Infarction (LIBERATE Study - Phase 2b)

Recognizing the potential broader applicability of GPVI inhibition in acute thrombotic events, Acticor Biotech extended the clinical development of glenzocimab to include ST-segment elevation myocardial infarction (STEMI).[3] The LIBERATE study, a Phase 2b clinical trial, was initiated as an academic partnership with the University of Birmingham in the UK to evaluate glenzocimab in the acute phase of STEMI.[3] The primary objective of the LIBERATE trial is to assess the efficacy of glenzocimab in reducing myocardial infarct size, measured at Day 90 post-treatment. Infarct size is a critical determinant of long-term cardiac function and prognosis following MI.[3] The final results from the LIBERATE study were anticipated in the fourth quarter of 2025 [13]; however, the liquidation of Acticor Biotech introduces uncertainty regarding the completion, full analysis, and public dissemination of these trial results.

B. Pulmonary Embolism (BREATHE Study)

Acticor Biotech had also considered the development of glenzocimab for the treatment of pulmonary embolism (PE). A Phase 2 study, codenamed BREATHE, was planned for this indication.[17] However, the company made a strategic decision to postpone the launch of the BREATHE study. This decision was made to allow Acticor Biotech to concentrate its resources and efforts on the ongoing development programs in acute ischemic stroke and myocardial infarction.[17]

The expansion of glenzocimab's development into myocardial infarction, as evidenced by the LIBERATE trial [3], suggests a belief in the fundamental role of GPVI-mediated platelet activation in a range of acute thrombotic cardiovascular conditions. Both AIS and acute MI are characterized by acute thrombus formation where platelets are key pathological players. If GPVI inhibition proves effective and safe in one such setting, it is plausible that it could offer benefits in others. However, it is important to note that the specific vascular environments, hemodynamics, and standard concomitant therapies differ significantly between these conditions, which can influence treatment outcomes. The potential of glenzocimab to reduce infarct size in MI, if demonstrated, would be a clinically significant finding.

The decision to postpone the pulmonary embolism study (BREATHE) [17] likely reflected a strategic prioritization of resources by Acticor Biotech, even before the company faced more acute financial distress. Developing a novel therapeutic agent across multiple major indications concurrently is an extremely resource-intensive endeavor. Pharmaceutical companies often make strategic choices to focus on indications with the perceived strongest preclinical rationale, highest unmet medical need, largest market potential, or highest probability of clinical success, given their available funding and capabilities. Prioritizing stroke and MI, which are major causes of morbidity and mortality with established (though still improvable) clinical development pathways for antithrombotics, over PE might have been an early indication of the company's efforts to manage its development pipeline within its financial constraints.

The future of the LIBERATE trial and the availability of its results are now uncertain due to Acticor Biotech's liquidation. Should the trial data become available, its findings could be pivotal in determining any potential future for the glenzocimab asset. Positive results in MI could potentially rekindle interest from other pharmaceutical entities, even if the broader stroke indication proved challenging, as different thrombotic conditions may exhibit differential responses to the same therapeutic mechanism.

VII. Safety and Tolerability Profile

A. Overview of Adverse Events Across Clinical Trials

Glenzocimab has been administered to a considerable number of subjects throughout its clinical development program. As of July 2024, over 400 patients had received active doses of glenzocimab (typically 1000 mg), and in total, more than 800 subjects had been included in clinical trials involving the drug.[2] Across these studies, particularly in the context of acute ischemic stroke, glenzocimab has generally been reported to have a favorable safety profile and to be well-tolerated, without major unexpected safety concerns emerging.[2]

B. Focus on Bleeding Risk, Including Intracranial Hemorrhage (ICH)

A central tenet of glenzocimab's development was its potential to offer antithrombotic efficacy with a reduced risk of bleeding complications, a critical consideration for any antiplatelet agent used in AIS.[2]

Data from the Phase 1b/2a ACTIMIS study provided initial supportive evidence for this favorable bleeding profile. In the Phase 2a portion, patients receiving the 1000 mg dose of glenzocimab experienced no symptomatic intracranial hemorrhage (sICH) (0%), compared to a 10% incidence (5 patients) in the placebo group.6 Furthermore, the occurrence of non-symptomatic hemorrhagic transformation was numerically lower in the glenzocimab group (31%) than in the placebo group (50%).6

The subsequent, larger Phase 2/3 ACTISAVE trial reaffirmed the favorable safety profile of glenzocimab, with no new safety signals identified.11 While detailed comparative rates for specific types of ICH from ACTISAVE were not as prominently featured in the available summaries as those from ACTIMIS, the consistent message was one of good overall tolerability and safety.

This clinical safety profile is further supported by preclinical studies in mouse models of ICH, which indicated that glenzocimab did not worsen bleeding severity. This contrasted with the GPIIb/IIIa inhibitor eptifibatide, which significantly increased bleeding in the same models.9

C. Safety in Conjunction with Standard Stroke Therapies

Glenzocimab was primarily evaluated as an adjunctive therapy, administered in addition to the standard of care for AIS. This standard care included intravenous thrombolysis with alteplase and/or mechanical thrombectomy.[2] The safety profile of glenzocimab appeared to be maintained when used in these combination settings, without evidence of synergistic toxicity or a significant increase in treatment-related adverse events beyond what might be expected from the standard therapies alone.

Table 3: Comparative Safety Data (Glenzocimab 1000mg vs. Placebo) for Key Adverse Events from ACTIMIS (Phase 2a)

Adverse EventGlenzocimab 1000mg (N=53) N (%)Placebo (N=53) N (%)Reference(s)
Symptomatic ICH0 (0%)5 (10%)6
Non-symptomatic HT17 (31%)26 (50%)6
All-Cause Mortality4 (7%)11 (21%)6
Serious Adverse Events (SAEs)Data not specified in snippetsData not specified
TEAEs leading to discontinuationData not specified in snippetsData not specified
Note: Specific comparative rates for SAEs and discontinuations from ACTISAVE were not detailed in the provided snippets. HT = Hemorrhagic Transformation.

The most consistent and arguably most significant positive signal emerging from the glenzocimab clinical development program is its safety profile, particularly the observed low risk of symptomatic ICH, even when administered concomitantly with thrombolytic agents and/or during mechanical thrombectomy procedures.[6] The results from the ACTIMIS Phase 2a study, showing 0% sICH in the glenzocimab arm versus 10% in the placebo arm [6], are particularly noteworthy. Although the larger ACTISAVE trial did not report such a dramatic difference in sICH (or these specific data points were not highlighted in the available summaries), the overall message of a favorable safety profile was maintained. This inherently low bleeding propensity is a crucial potential advantage over many existing or developmental potent antiplatelet or anticoagulant strategies in the acute stroke setting.

The preclinical data demonstrating no exacerbation of bleeding in established ICH models [9] lend additional credence to the clinical observations regarding glenzocimab's bleeding safety profile. Such concordance between preclinical bleeding models and clinical safety findings strengthens confidence in the drug's intrinsic properties. If glenzocimab genuinely does not worsen existing bleeds, this would be a unique and highly valuable characteristic among antithrombotic agents.

Even if broad efficacy remains unproven in heterogeneous patient populations, this favorable safety profile, particularly concerning bleeding, might still render glenzocimab (or the GPVI target itself) an interesting candidate for future research. This could be in very specific, high-bleeding-risk patient populations or in combination with other therapies known to increase bleeding risk, where glenzocimab's safety could be an enabling factor for more aggressive treatment regimens.

VIII. Regulatory Status and Designations

A. European Medicines Agency (EMA)

In July 2022, glenzocimab was granted Priority Medicines (PRIME) status by the European Medicines Agency (EMA) for the treatment of stroke.[2] The PRIME scheme is designed to enhance support for the development of medicines that target an unmet medical need. This designation is awarded to medicines that may offer a major therapeutic advantage over existing treatments, or benefit patients with no treatment options. It facilitates early and enhanced interaction and dialogue with EMA, aiming to optimize development plans and potentially accelerate the assessment of a marketing authorization application.[2] Furthermore, the clinical trial design of the ACTISAVE study had received scientific advice from the EMA, indicating its potential suitability to support a future marketing authorization application, contingent upon the generation of positive and robust clinical data.[19]

B. U.S. Food and Drug Administration (FDA)

Acticor Biotech had engaged in regulatory discussions with the U.S. Food and Drug Administration (FDA) regarding the development of glenzocimab. Plans were in place for a Type C meeting in early 2023. The primary objective of this meeting was to seek FDA feedback on the design of the ACTISAVE study and its adequacy to support a potential future Biologics License Application (BLA) for glenzocimab in the United States.[19]

A request for Fast Track designation was submitted to the FDA on October 11, 2022. This submission was based on the promising results from the Phase 1b/2a ACTIMIS study.[19] The FDA acknowledged that acute ischemic stroke is a serious condition and recognized that glenzocimab's development program was designed to address a serious aspect of this condition. However, the agency determined that additional clinical evidence, presumably from the then-ongoing ACTISAVE study, was required before Fast Track designation could be granted. Acticor Biotech was encouraged to resubmit the Fast Track request with further supportive data from the ACTISAVE study.[19] The available information does not indicate that Fast Track designation was ultimately granted by the FDA.

The EMA's decision to grant PRIME status [2] represented a significant early regulatory endorsement for glenzocimab. This suggested that European regulatory authorities perceived substantial therapeutic innovation and a potential to address an unmet medical need, based on the drug's novel mechanism of action and the initial clinical data emerging from the ACTIMIS trial. Such a designation is not easily obtained and typically signals strong regulatory interest, which would have been an encouraging development for Acticor Biotech and its stakeholders, potentially paving the way for a smoother regulatory pathway in Europe had the subsequent pivotal trial results been unequivocally positive.

In contrast, the FDA's more cautious approach regarding Fast Track designation, stipulating the need for additional data from the ACTISAVE trial [19], foreshadowed the higher bar for demonstrating efficacy that ultimately proved challenging for glenzocimab in its larger clinical trials. While safety and a plausible mechanistic rationale are important, the FDA often places a strong emphasis on clear and robust evidence of clinical benefit before granting expedited pathway designations. The ACTIMIS results, though promising in terms of safety and showing some positive trends, were likely deemed insufficient by the FDA to fully support the efficacy claims necessary for Fast Track designation without further validation from the larger, more rigorously designed ACTISAVE study. This stance, in retrospect, aligned with the eventual outcome of ACTISAVE not meeting its primary efficacy endpoint.

The subsequent failures of the ACTISAVE and GREEN trials to meet their primary efficacy endpoints significantly diminish the immediate impact of the earlier PRIME designation and fundamentally alter any potential BLA pathway. Regulatory strategies are contingent upon successful clinical trial outcomes. With these setbacks, any future regulatory engagement by a new entity, should the glenzocimab asset be acquired, would necessitate a complete re-evaluation, likely focusing on very specific patient subpopulations or requiring entirely new pivotal studies based on revised clinical hypotheses and endpoints.

IX. Developer Profile and Current Status

A. Acticor Biotech

Acticor Biotech was a clinical-stage biopharmaceutical company founded in 2013. It originated as a spin-off from the esteemed French National Institute of Health and Medical Research (INSERM).[2] The company's primary strategic focus was the research and development of its lead innovative drug candidate, glenzocimab (ACT017). The development efforts were concentrated on addressing cardiovascular emergencies, with a particular and significant emphasis on the treatment of acute ischemic stroke.[2] Acticor Biotech had achieved public listing status, with its shares traded on the Euronext Growth Paris market since November 2021.[20]

B. Recent Corporate Developments (Receivership and Liquidation)

The trajectory of Acticor Biotech took a significant downturn following disappointing results from its key clinical trials. The announcement in April 2024 that the pivotal Phase 2/3 ACTISAVE trial for glenzocimab in AIS failed to meet its primary efficacy endpoint was a major setback.[11] This was compounded by the subsequent recommendation in July 2024 to discontinue the Phase 2/3 GREEN trial, also in AIS, due to futility based on an interim analysis.[13]

These clinical trial failures had severe financial repercussions for the company. As of April/May 2024, Acticor Biotech had indicated that its existing cash reserves would only be sufficient to fund its operations until October 2024.[11] Consequently, on August 6, 2024, the Paris Commercial Court officially opened receivership proceedings (redressement judiciaire) for Acticor Biotech.[21] In an attempt to salvage value from its primary asset, an open call for tenders was launched in September 2024 as part of the receivership process, seeking a continuation or sale plan for glenzocimab and related assets.[20]

Despite these efforts, the court-appointed administrator requested the conversion of the receivership into liquidation proceedings in December 2024.[20] This request was granted, and on January 2, 2025, the Paris Commercial Court formally declared Acticor Biotech in liquidation proceedings.[3] Following this decision, a request for the delisting of Acticor Biotech shares from the Euronext Growth Paris market was anticipated.[3]

C. Implications for Glenzocimab's Future

With the liquidation of Acticor Biotech, the future of glenzocimab is uncertain. The company's primary assets, including the intellectual property rights associated with glenzocimab (reportedly comprising three patent families, with the first set to expire in 2036) [3], and the substantial preclinical and clinical data package (generated from studies involving over 800 subjects, with more than 400 directly exposed to glenzocimab) [3], will now be subject to the liquidation process. The continued clinical development of glenzocimab hinges entirely on the possibility of these assets being acquired by another pharmaceutical or biotechnology entity. Such an acquirer would need to perceive continued value in the drug candidate and be willing to undertake the significant investment required for further, likely more targeted, clinical investigations.[20]

The sequence of events, from disappointing late-stage trial results [11] to financial instability [11] and culminating in corporate liquidation [3], illustrates a direct causal chain frequently observed in the biopharmaceutical industry. Clinical-stage biotech companies, particularly those heavily reliant on the success of a single lead asset, are highly vulnerable to the outcomes of pivotal clinical trials. Negative data can severely erode investor confidence, impede the ability to raise further capital, and potentially trigger financial defaults if funding is tied to specific development milestones.

Despite Acticor's demise, the glenzocimab asset package itself may still hold residual value. The extensive safety database, consistently highlighting a low bleeding risk, is a significant de-risking component for any future development efforts. The drug's unique mechanism of action targeting GPVI also remains scientifically intriguing. If a new entity can identify a scientifically sound and commercially viable path forward – perhaps by focusing on the specific patient subgroups that showed positive trends in the ACTISAVE post-hoc analyses [11], or by exploring alternative indications such as myocardial infarction (pending results from the LIBERATE trial [3]) – the asset could potentially be acquired at a valuation reflecting its current developmental stage and associated risks. The reported mortality reduction in a small subgroup of intracerebral hemorrhage patients from one trial [3], while likely hypothesis-generating due to small numbers, might also attract niche research interest.

The story of glenzocimab and Acticor Biotech serves as a stark reminder of the inherent risks and high attrition rates in drug development, particularly for complex and challenging indications like acute ischemic stroke. This experience may temper investor enthusiasm for other early-stage stroke assets with novel mechanisms unless they can demonstrate exceptionally robust and early efficacy signals, coupled with strong translational science.

X. Discussion and Future Perspectives

A. Synthesis of Key Findings

Glenzocimab, a humanized Fab fragment targeting the platelet receptor GPVI, was developed with the aim of providing a novel antiplatelet therapy for acute ischemic stroke, distinguished by its potential for efficacy with an improved safety profile, specifically a reduced risk of bleeding complications. Its mechanism of action involves the inhibition of GPVI-mediated platelet aggregation and thrombus formation through the prevention of GPVI dimerization and its subsequent interaction with key ligands such as collagen and fibrin.[1]

The clinical development program for glenzocimab in AIS yielded mixed results. Early-phase studies, notably the Phase 1b/2a ACTIMIS trial, demonstrated a favorable safety profile for the 1000 mg dose, including a striking reduction in symptomatic intracranial hemorrhage and overall mortality when compared to placebo.[2] These promising initial findings, however, were not replicated in terms of primary efficacy in the larger, pivotal Phase 2/3 ACTISAVE trial, which failed to show a significant reduction in severe disability or death (mRS 4-6) in the broad AIS population studied.[11] Despite this, intriguing trends towards benefit in achieving an excellent functional outcome (mRS 0-1) were observed in specific patient subgroups within ACTISAVE, particularly those who achieved complete recanalization following mechanical thrombectomy.[11] The adjunctive GREEN trial, focused on patients undergoing mechanical thrombectomy, was subsequently halted prematurely due to futility.[13] Throughout its clinical evaluation, glenzocimab consistently maintained a favorable safety profile, with low rates of sICH being a persistent positive attribute.[6] The developing company, Acticor Biotech, has since entered liquidation proceedings, leaving the immediate future development of glenzocimab uncertain.[3]

B. Unmet Needs and Potential Future Role of Glenzocimab

A significant unmet medical need persists for safer and more effective adjunctive therapies in the management of AIS. Current treatments, while beneficial, have limitations, and improvements in neurological outcomes are still urgently sought. Despite the setbacks in the broad AIS population, the data from the glenzocimab program, particularly the consistent safety profile and the efficacy signals in specific subgroups (e.g., mRS 0-1 trend in eTICI 3 recanalized patients from ACTISAVE [11]), might warrant consideration for highly targeted future trials, should a new entity acquire the asset. The potential utility of glenzocimab in other acute thrombotic indications, such as myocardial infarction (as investigated in the LIBERATE study [3]), or the exploratory finding of reduced mortality in a small cohort of ICH patients [3], could also represent avenues for future exploration, although these would require substantial further investigation and robust data generation.

C. Challenges and Opportunities for Future Development/Asset Utilization

The path forward for glenzocimab, or any therapeutic based on its technology, faces considerable challenges. These include the need to overcome the negative perception arising from the failure of large pivotal trials, the difficulty in securing substantial funding for further development, especially in a post-liquidation context, and the complexities of designing and executing clinical trials in highly specific and potentially small stroke subpopulations. Such trials can be lengthy and costly to recruit for.

However, opportunities may also exist. The extensive existing safety database for glenzocimab [3] is a valuable asset that de-risks certain aspects of future development. The unique mechanism of action targeting GPVI [1] remains scientifically compelling and might yet hold promise if the optimal patient population or a synergistic combination therapy approach can be precisely identified. The intellectual property portfolio associated with glenzocimab [3] could provide a period of market exclusivity if development were to be successfully revived.

The journey of glenzocimab exemplifies the "valley of death" often encountered in drug development, where promising early-phase data and a strong mechanistic rationale do not invariably translate into late-stage clinical success. This is particularly true for complex and heterogeneous conditions such as acute ischemic stroke. The interplay between scientific potential, the rigors of clinical trial execution, definitive trial outcomes, and the financial viability of the developing company is starkly illustrated by this case.

The primary value of the glenzocimab asset, following Acticor Biotech's liquidation, likely resides in its comprehensive safety database and the potential for a highly targeted therapeutic approach. This could involve focusing on a very specific AIS subgroup where the mechanistic rationale aligns with observed trends (such as the mRS 0-1 benefit in fully recanalized patients [11]) or perhaps exploring an alternative, less heterogeneous thrombotic indication. Any new entity undertaking such a development would require a compelling, data-driven rationale and a cost-effective strategy to meticulously de-risk further clinical investigation. The observed reduction in mortality within a specific ICH subgroup [3], while intriguing, is likely too preliminary and would necessitate substantial foundational research before any clinical pursuit could be justified.

XI. Conclusion

Glenzocimab (ACT017) is a humanized Fab fragment targeting platelet GPVI, developed as a novel antiplatelet agent for acute ischemic stroke. Its mechanism, involving the inhibition of GPVI dimerization and its interaction with collagen and fibrin, offered a promising approach to reducing thrombosis with a potentially lower bleeding risk. Preclinical studies supported this profile, showing antithrombotic efficacy without significantly increasing bleeding.

Early clinical development (Phase 1b/2a ACTIMIS trial) was encouraging, demonstrating a favorable safety profile, notably a reduction in symptomatic intracranial hemorrhage and mortality in glenzocimab-treated patients compared to placebo. However, these positive signals did not translate into success in the larger Phase 2/3 ACTISAVE trial, which failed to meet its primary efficacy endpoint of reducing severe disability or death (mRS 4-6) in the overall AIS population. While trends towards benefit in achieving excellent functional outcomes (mRS 0-1) were noted in certain subgroups, particularly patients with complete recanalization after thrombectomy, these were not sufficient to demonstrate overall efficacy. The subsequent GREEN trial, focusing on patients undergoing mechanical thrombectomy, was halted for futility.

Despite the efficacy setbacks, glenzocimab consistently demonstrated a favorable safety profile across studies. The developing company, Acticor Biotech, entered liquidation proceedings in early 2025, largely as a consequence of these clinical trial outcomes. The future of glenzocimab now depends on the potential acquisition and further development of its assets by another entity. Any such endeavor would likely need to focus on highly specific patient populations where the drug's mechanism and observed trends might offer a viable therapeutic benefit, or explore alternative indications, leveraging the existing extensive safety data. The story of glenzocimab underscores the significant challenges in developing effective and safe treatments for acute ischemic stroke.

Works cited

  1. Population Pharmacokinetic/Pharmacodynamic Modeling of Glenzocimab (ACT017) a Glycoprotein VI Inhibitor of Collagen‐Induced Platelet Aggregation - PubMed Central, accessed June 13, 2025, https://pmc.ncbi.nlm.nih.gov/articles/PMC7496554/
  2. About Us - Acticor Biotech, accessed June 13, 2025, https://www.acticor-biotech.com/company/about-us
  3. Acticor Biotech announces its liquidation proceedings - Business Wire, accessed June 13, 2025, https://www.businesswire.com/news/home/20250105406020/en/Acticor-Biotech-announces-its-liquidation-proceedings
  4. Targeting platelet GPVI with glenzocimab: a novel mechanism for inhibition - PMC, accessed June 13, 2025, https://pmc.ncbi.nlm.nih.gov/articles/PMC10119634/
  5. Glenzocimab (Anti-GP6 / Glycoprotein-6) | Read Reviews & Product Use Citations, accessed June 13, 2025, https://www.selleckchem.com/products/glenzocimab-anti-gp6-glycoprotein-6.html
  6. Safety and efficacy of platelet glycoprotein VI inhibition in acute ischaemic stroke (ACTIMIS): a randomised, double-blind, placebo-controlled, phase 1b/2a trial - PubMed, accessed June 13, 2025, https://pubmed.ncbi.nlm.nih.gov/38267188/
  7. Efficacy and safety study of glenzocimab as add on therapy in AIS, accessed June 13, 2025, https://www.hra.nhs.uk/planning-and-improving-research/application-summaries/research-summaries/efficacy-and-safety-study-of-glenzocimab-as-add-on-therapy-in-ais/
  8. Glenzocimab in acute ischemic stroke - Acticor Biotech, accessed June 13, 2025, https://www.acticor-biotech.com/our-product/glenzocimab-in-acute-ischemic-stroke
  9. Comparative Effects of Glenzocimab and Eptifibatide on Bleeding Severity in 2 Mouse Models of Intracranial Hemorrhage - PubMed Central, accessed June 13, 2025, https://pmc.ncbi.nlm.nih.gov/articles/PMC12074769/
  10. (PDF) Comparative Effects of Glenzocimab and Eptifibatide on Bleeding Severity in 2 Mouse Models of Intracranial Hemorrhage - ResearchGate, accessed June 13, 2025, https://www.researchgate.net/publication/388121575_Comparative_Effects_of_Glenzocimab_and_Eptifibatide_on_Bleeding_Severity_in_2_Mouse_Models_of_Intracranial_Hemorrhage
  11. ACTICOR BIOTECH: Presentation of the Main Results of the Phase 2/3 ACTISAVE Study in the Treatment of Stroke at ESOC 2024 - Euronext Markets, accessed June 13, 2025, https://live.euronext.com/en/products/equities/company-news/2024-05-15-acticor-biotech-presentation-main-results-phase-23
  12. ACTICOR BIOTECH: Presentation of the Main Results of the Phase 2/3 ACTISAVE Study in the Treatment of Stroke at ESOC 2024 - Business Wire, accessed June 13, 2025, https://www.businesswire.com/news/home/20240515314114/en/ACTICOR-BIOTECH-Presentation-of-the-Main-Results-of-the-Phase-23-ACTISAVE-Study-in-the-Treatment-of-Stroke-at-ESOC-2024
  13. Acticor Biotech Announces the Conclusions of the Interim Futility Analysis in the GREEN Study - Business Wire, accessed June 13, 2025, https://www.businesswire.com/news/home/20240725439354/en/Acticor-Biotech-Announces-the-Conclusions-of-the-Interim-Futility-Analysis-in-the-GREEN-Study
  14. Acticor Biotech updates its glenzocimab clinical development plan for the treatment of cardiovascular emergencies, accessed June 13, 2025, https://ml-eu.globenewswire.com/Resource/Download/98d5fa92-9757-4a4c-b697-76632ac4dfe2
  15. ESOC 2024: Second Announcement of Large Clinical Trials - European Stroke Organisation, accessed June 13, 2025, https://eso-stroke.org/wp-content/uploads/Curtain-Raiser-2.pdf
  16. ESOC 2024: first large clinical trials announced. - European Stroke Organisation, accessed June 13, 2025, https://eso-stroke.org/wp-content/uploads/Curtain-Raiser-1.pdf
  17. Update on Acticor Biotech's clinical developments with glenzocimab in the treatment of cardiovascular emergencies - Business Wire, accessed June 13, 2025, https://www.businesswire.com/news/home/20230109005745/en/Update-on-Acticor-Biotechs-clinical-developments-with-glenzocimab-in-the-treatment-of-cardiovascular-emergencies
  18. Acticor Biotech announces its liquidation proceedings - Euronext Markets, accessed June 13, 2025, https://live.euronext.com/en/products/equities/company-news/2025-01-06-acticor-biotech-announces-its-liquidation-proceedings
  19. Acticor Biotech Provides an Update on the Us Regulatory Discussion with the FDA, accessed June 13, 2025, https://www.businesswire.com/news/home/20221222005451/en/Acticor-Biotech-Provides-an-Update-on-the-Us-Regulatory-Discussion-with-the-FDA
  20. Acticor Biotech Liquidation Proceedings Announced - The Clinical Trial Vanguard, accessed June 13, 2025, https://www.clinicaltrialvanguard.com/news/acticor-biotech-liquidation-proceedings-announced/
  21. Acticor Biotech Launches a Call for Tenders to Provide a Continuation or Sale Plan as Part of the Receivership Procedure - Business Wire, accessed June 13, 2025, https://www.businesswire.com/news/home/20240912684231/en/Acticor-Biotech-Launches-a-Call-for-Tenders-to-Provide-a-Continuation-or-Sale-Plan-as-Part-of-the-Receivership-Procedure
  22. Acticor Biotech Announces that the Court Judgment Has Been Postponed Until January 2, 2025 - Business Wire, accessed June 13, 2025, https://www.businesswire.com/news/home/20241219912586/en/Acticor-Biotech-Announces-that-the-Court-Judgment-Has-Been-Postponed-Until-January-2-2025
  23. Press Releases - Acticor Biotech, accessed June 13, 2025, https://www.acticor-biotech.com/press-releases
  24. Acticor Biotech Strengthens Its Financial Structure - Business Wire, accessed June 13, 2025, https://www.businesswire.com/news/home/20221017005919/en/Acticor-Biotech-Strengthens-Its-Financial-Structure

Published at: June 13, 2025

This report is continuously updated as new research emerges.

MedPath

Empowering clinical research with data-driven insights and AI-powered tools.

© 2025 MedPath, Inc. All rights reserved.